EP4117658A1 - Substituted isoxazoles as selective nav1.7 inhibitors for pain treatment and method of pain treatment - Google Patents

Substituted isoxazoles as selective nav1.7 inhibitors for pain treatment and method of pain treatment

Info

Publication number
EP4117658A1
EP4117658A1 EP21767873.9A EP21767873A EP4117658A1 EP 4117658 A1 EP4117658 A1 EP 4117658A1 EP 21767873 A EP21767873 A EP 21767873A EP 4117658 A1 EP4117658 A1 EP 4117658A1
Authority
EP
European Patent Office
Prior art keywords
navi
group
phenyl
inhibitor
selectivity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21767873.9A
Other languages
German (de)
French (fr)
Other versions
EP4117658A4 (en
Inventor
Doug Franz
Sara DIBRELL
Robynne NEFF
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Texas System
Original Assignee
University of Texas System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Texas System filed Critical University of Texas System
Publication of EP4117658A1 publication Critical patent/EP4117658A1/en
Publication of EP4117658A4 publication Critical patent/EP4117658A4/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/08Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/10Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D261/18Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/20Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • opioid overdose including both prescription and illicit opioids.
  • prescription opioids can be used to treat moderate-to-severe pain and are often prescribed following surgery or injury, or for health conditions such as cancer.
  • prescription opioids for the treatment of chronic, non-cancer pain, such as back pain or osteoarthritis, despite serious risks and the lack of evidence about their long-term effectiveness.”
  • More than 191 million opioid prescriptions were dispensed to patients in the United States in 2017.
  • Opioids are addictive and patients taking them develop tolerance to their pharmacologic benefits, necessitating increased dosages.
  • Substituted isoxazoles are potent inhibitors of Navi.7 and their structure-activity relationships have been found tunable for selectivity over Navi .5.
  • Structure-activity relationship (SAR) studies demonstrated that subtype selectivity (Navi.7 vs. Navi.5) could be improved with methylation of the amide nitrogen or ortho-substitution on the phenyl ring in the 5-position.
  • FIG. 1 illustrates a restricted rotation axis of chirality of telenzepine as is known in the art.
  • FIG. 2 illustrates the core structure of the inventive tri-substituted isoxazole compound in accordance with the present disclosure.
  • FIG. 3 is a graph of the results from a mouse automated formalin test an isoxazole compound in accordance with the present disclosure.
  • FIG. 4 is a density function theory (B3LYP/6-31G) geometry minimization of a simplified methyl ester analog of compound in accordance with the present invention.
  • FIG. 5 is a pharmacophore model and SAR on the tri-substituted isoxazole Navi.7 inhibitors in accordance with the present disclosure.
  • FIG. 6 is a synthetic pathway for atropisomeric 3, 4, 5 tri-substituted isoxazoles employed for SAR analysis in Navl.7/Navl.5 in vitro assays in accordance with the present disclosure.
  • a new class of isoxazole-based Navi.7 inhibitors are disclosed that demonstrate potent inhibition of hNavl.7, tunable selectivity over hNavl.5 and possess ideal starting physiochemical properties for further drug development.
  • SAR structure-activity relationship
  • a previously unrecognized structural feature in this class of compounds, atropisomerism has been identified.
  • the dramatic role that chirality and three-dimensionality plays in influencing biological activity is well established.
  • atropisomerism an example of axial chirality, has never been exploited in SAR studies of isoxazole-based small molecules despite their role as a privileged scaffold in medicinal chemistry.
  • telenzepine A well-known industrial example of differential activity due to atropisomerism is telenzepine, shown in Fig. 1. Telenzipine is a selective muscarinic antagonist for the treatment of peptic ulcers. The (-)-isomer was eventually found to be 500x less active with much less selectivity than the (+)-isomer at muscarinic receptors in the rat cerebral cortex. Unfortunately, strategies to address atropisomerism in drug discovery are lacking and there are currently no specific guidelines from regulatory agencies on how to handle this time-dependent chirality despite its prevalence and impact on the pharmaceutical industry.
  • the present invention exploits the previously unrecognized form of atropisomerism in 3, 4, 5 -tri substituted isoxazoles to yield a new class of Navi .7 inhibitors.
  • This provides opportunities for multiple discoveries with respect to the synthetic design of atropisomeric isoxazoles, their characterization and kinetics of chirality, and the subsequent understanding of how this chirality element affects Navi.7 potency and selectivity over Navi.5. More broadly, given the prevalence of substituted isoxazoles in numerous drug discovery programs, which will aid in designing new strategies to recognize and understand the positive or negative impact of atropisomerism across multiple therapeutic programs.
  • the 3, 4, 5 -tri substituted isoxazole in accordance with the present invention is based upon general formula I, including a stereoisomer, enantiomer, atropisomer, mixture of enantiomers, mixture of diastereomers, mixture of atropisomers, or isotopic variant thereof; or a pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof:
  • R1 is selected from the group of methyl, phenyl, chlorophenyl, dichlorophenyl, fluorophenyl, trifluoromethyl, methoxypehnyl, cyanophenyl, pyridine, furan, and thiophene, and combinations thereof;
  • R2 is selected from the group of hydrogen, methyl, trifluoromethyl, halogen, alkynyl, phenyl, amide, methylphenyl, and fluoromethylphenyl, and combinations thereof;
  • R3 is selected from the group of hydrogen, keto, thioketo, and combinations thereof;
  • R4 is selected from the group of 1-10 carbon branched or straight chain alkyl, hydroxyalky, cyclic, heterocyclic, sulfide, aldehyde, phenyl, and combinations thereof; and
  • R5 is selected from the group of hydrogen, methyl, and saturated or unsaturated cycloalkanes having 3-6 carbon atoms.
  • Compound 124 includes a meta-substitution on the phenyl ring in the 5-position that results in reversing subtype selectivity.
  • Compound 124 was found to be very potent againstNavl .5 (36 nM) as well as Navi.7 (120 nM) again, supporting that subtle structural differences have a profound influence on both potency and selectivity.
  • Navi.7 potency was realized when a substitution was introduced at the 4-position of the isoxazole ring as exemplified by the 4-phenyl substitution in compound 106.
  • Compound 100 was administered using the mouse automated formalin model at a dosage of 135 mg/kg p.o. Dosage was selected based upon projection from mouse exposure at 5 mg/kg. Early phase and late phase tonic events were measured with a Tmax equal to 15 minutes and a Tl/2 equal to 1.1 hours. Table 2, below, and FIG. 3 summarize the results of the test demonstrating the Navi.7 inhibitory activity of compound 100 relative to vehicle.
  • FIG. 5 outlines the synthesis of 3,4,5-trisubstituted isoxazoles in accordance with the present invention.
  • Compound 106 was synthesized by this synthetic route using phenyl boronic acid.
  • atropisomers around the C-C bond at the 4-position are contemplated with either a single ortho- or meta-substituent.
  • Atropisomeric isoxazoles with simple ortho-substitution from aryl boronic acids that includes ortho-fluorine, -chlorine, -bromine, -methyl, and -methoxy may also be made by this synthetic route.
  • Chiral separation of tri substituted atropisomeric isoxazoles may be accomplished by interrupting the synthetic route after the Suzuki cross-coupling reaction to yield two complementary methods for separating the atropisomers as shown in FIG. 5.
  • the methyl ester offers an ideal site for chiral semi-preparative HPLC separation.
  • hydrolysis of the methyl ester to the corresponding carboxylic acid as a handle for separation via classical resolution.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)

Abstract

Tri-substituted isoxazole compounds and compound synthesis is disclosed. Tri-substituted isoxazole compounds having sodium gate channel Nav7 selectivity that are tunable for selectivity over Nav1.5. In particular, structure-activity relationship (SAR) studies demonstrated that subtype selectivity (Nav1.7 vs. Nav1.5) is improved with methylation of the amide nitrogen or ortho-substitution on the phenyl ring in the 5-position.

Description

Title:
[0001] SUBSTITUTED ISOXAZOLES AS SELECTIVE NAV1.7 INHIBITORS FOR PAIN TREATMENT AND METHOD OF PAIN TREATMENT
Background of the Invention
[0002] The annual cost of chronic pain has been estimated as being as high as $635 billion, which is greater than the combined yearly costs for cancer, heart disease and cancer. According to the Medical Expenditure Panel Survey in 2008, about 100 million adults in the United States were affected by chronic pain. Chronic pain limits suffers functional status and adversely impacts their quality of life. Pain also complicates medical care for other ailments.
[0003] To further complicate the problem, from 1999-2017 almost 400,000 people have died from opioid overdose, including both prescription and illicit opioids. According to the Centers for Disease Control and Prevention “prescription opioids can be used to treat moderate-to-severe pain and are often prescribed following surgery or injury, or for health conditions such as cancer. In recent years, there has been a dramatic increase in the acceptance and use of prescription opioids for the treatment of chronic, non-cancer pain, such as back pain or osteoarthritis, despite serious risks and the lack of evidence about their long-term effectiveness.” More than 191 million opioid prescriptions were dispensed to patients in the United States in 2017. Opioids are addictive and patients taking them develop tolerance to their pharmacologic benefits, necessitating increased dosages. Moreover, opioid administration frequently carries with it deleterious side effects which also limits their long-term use and effectiveness. Unfortunately, chronic pain continues to expand as an overwhelming burden to the health care system while current therapies suffer from limitations in efficacy, undesirable side effects and contribute to a growing drug addiction crisis in the U.S.
[0004] Accordingly, identifying alternatives for chronic pain treatment has become a high priority in the pain therapy and management field. Presented in this application are substituted isoxazole compounds having antagonistic activity at the Navi.7 voltage-gated sodium channel.
[0005] Recent studies have suggested that most chronic pain states are maintained by persistent peripheral nociceptive triggers. As a result, the voltage-gated sodium channel Navi.7 has attracted the greatest attention as an alternative pharmacological target due to strong genetic validation implicating its pivotal role in pain perception in the peripheral nervous system. Voltage-gated sodium channels are essential for electrogenesis in nerve cells. Genetic, structural and functional studies have shown that Navi .7 regulates sensory neuron excitability and is a major contributor to several sensory modalities including human pain disorders. Other indications include sense of smell, cough reflex, and epilepsy. The role of Navi.7 in acquired and inherited pain conditions, including chronic pain, has been well established. Small molecules that act as antagonists (synonymously, inhibitors) of Navi.7 have been shown to be potential therapeutic agents for the treatment of chronic pain in humans.
[0006] Despite intense effort, however, small molecule inhibitors of Navi.7 have yet to recapitulate the powerful analgesic phenotype observed in Navi.7-null subjects shedding doubt on the druggability of this target. This lack of efficacy has, in part, been attributed to poor isoform selectivity of all reported inhibitors over other sodium channel subtypes, which elicits dose- limiting side effects. In particular, tuning out inhibition of the cardiac Navi .5 subtype, which could lead to significant cardiovascular adverse events, has been a significant challenge to overcome.
Summary of the Invention
[0007] Substituted isoxazoles are potent inhibitors of Navi.7 and their structure-activity relationships have been found tunable for selectivity over Navi .5. Structure-activity relationship (SAR) studies demonstrated that subtype selectivity (Navi.7 vs. Navi.5) could be improved with methylation of the amide nitrogen or ortho-substitution on the phenyl ring in the 5-position.
[0008] It has been found that subtle structural differences have a profound influence on both IC50 potency and selectivity of Navi.7 over Navi.5.
[0009] It has been found that a 4-phenyl substitution increases Navi.7 inhibition, particularly, where the 4-phenyl substituent is substantially orthogonal to the isoxazole ring.
[0010] The structural activity relationships between different 3,4,5 substitutions on the isoxazole ring have revealed that atropisomerism around the carbon-carbon bond at the 4-position affords chirality that enhances Navi.7 inhibition and selectivity over Navi.5.
[0011] A synthesis of atropisomeric 3, 4, 5 -tri substituted isoxazole is also disclosed. Brief Description of the Drawings
[0012] FIG. 1 illustrates a restricted rotation axis of chirality of telenzepine as is known in the art. [0013] FIG. 2 illustrates the core structure of the inventive tri-substituted isoxazole compound in accordance with the present disclosure.
[0014] FIG. 3 is a graph of the results from a mouse automated formalin test an isoxazole compound in accordance with the present disclosure.
[0015] FIG. 4 is a density function theory (B3LYP/6-31G) geometry minimization of a simplified methyl ester analog of compound in accordance with the present invention.
[0016] FIG. 5 is a pharmacophore model and SAR on the tri-substituted isoxazole Navi.7 inhibitors in accordance with the present disclosure.
[0017] FIG. 6 is a synthetic pathway for atropisomeric 3, 4, 5 tri-substituted isoxazoles employed for SAR analysis in Navl.7/Navl.5 in vitro assays in accordance with the present disclosure.
Detailed Description of the Preferred Embodiments
[0018] A new class of isoxazole-based Navi.7 inhibitors are disclosed that demonstrate potent inhibition of hNavl.7, tunable selectivity over hNavl.5 and possess ideal starting physiochemical properties for further drug development. As part of the structure-activity relationship (SAR) identification, a previously unrecognized structural feature in this class of compounds, atropisomerism, has been identified. The dramatic role that chirality and three-dimensionality plays in influencing biological activity is well established. However, atropisomerism, an example of axial chirality, has never been exploited in SAR studies of isoxazole-based small molecules despite their role as a privileged scaffold in medicinal chemistry. This has led to identifying a potent Navi.7 inhibitor with high levels (>100x) of selectivity over Navi.5 that can serve as a tool compound to further elucidate isoform selectivity and provide the foundation for a drug discovery program for chronic pain treatment. Without intending to be bound to theory, we posit that this previously unknown atropisomerism will serve as a fine-tuning mechanism to not only increase potency but also function as a unique architectural handle to enhance Navi.7 selectivity over Navi.5.
[0019] Drug discovery is intensely affected by chirality. For the past 20 years, medicinal chemists have increasingly tried to mimic nature’s exquisite control of chirality into their synthetic drug discovery programs. The advantages of developing a single stereoisomer drug substance over mixtures (i.e., single enantiomer vs. racemate) are numerous and include improved efficacy, reduced off-target effects, and avoid stereospecific differences in pharmacokinetics and pharmacodynamics. However, an often-overlooked form of chirality in drug discovery is atropisomerism occurs due to a significant rotational barrier (~22 kcal/mol) about a rotationally stable bond axis that imparts asymmetry into the molecule and allows for the potential isolation of conformational stereoisomers. As with classical sources of chirality, these conformationally distinct stereoisomers can have significant differential biological profiles. A well-known industrial example of differential activity due to atropisomerism is telenzepine, shown in Fig. 1. Telenzipine is a selective muscarinic antagonist for the treatment of peptic ulcers. The (-)-isomer was eventually found to be 500x less active with much less selectivity than the (+)-isomer at muscarinic receptors in the rat cerebral cortex. Unfortunately, strategies to address atropisomerism in drug discovery are lacking and there are currently no specific guidelines from regulatory agencies on how to handle this time-dependent chirality despite its prevalence and impact on the pharmaceutical industry.
[0020] The present invention exploits the previously unrecognized form of atropisomerism in 3, 4, 5 -tri substituted isoxazoles to yield a new class of Navi .7 inhibitors. This provides opportunities for multiple discoveries with respect to the synthetic design of atropisomeric isoxazoles, their characterization and kinetics of chirality, and the subsequent understanding of how this chirality element affects Navi.7 potency and selectivity over Navi.5. More broadly, given the prevalence of substituted isoxazoles in numerous drug discovery programs, which will aid in designing new strategies to recognize and understand the positive or negative impact of atropisomerism across multiple therapeutic programs.
[0021] The 3, 4, 5 -tri substituted isoxazole in accordance with the present invention is based upon general formula I, including a stereoisomer, enantiomer, atropisomer, mixture of enantiomers, mixture of diastereomers, mixture of atropisomers, or isotopic variant thereof; or a pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof:
Wherein R1 is selected from the group of methyl, phenyl, chlorophenyl, dichlorophenyl, fluorophenyl, trifluoromethyl, methoxypehnyl, cyanophenyl, pyridine, furan, and thiophene, and combinations thereof;
Wherein R2 is selected from the group of hydrogen, methyl, trifluoromethyl, halogen, alkynyl, phenyl, amide, methylphenyl, and fluoromethylphenyl, and combinations thereof;
Wherein R3 is selected from the group of hydrogen, keto, thioketo, and combinations thereof; Wherein R4 is selected from the group of 1-10 carbon branched or straight chain alkyl, hydroxyalky, cyclic, heterocyclic, sulfide, aldehyde, phenyl, and combinations thereof; and Wherein R5 is selected from the group of hydrogen, methyl, and saturated or unsaturated cycloalkanes having 3-6 carbon atoms.
[0022] Specific 3, 4, 5 -tri substituted isoxazole compounds having Navi.7 inhibitor activity are listed in Table 1, below.
Table 1
[0023] As noted above, SAR studies have demonstrated that methylation of the amide nitrogen or ortho-substitution on the phenyl ring in the 5-position enhances sodium gate channel subtype selectivity (Navi .7 vs. Navi .5). Moreover, as is apparent from the enumerated 3,4,5-trisubstituted isoxazole compound structures 100-129 in Table 1, above, subtle structural differences have a profound influence on both IC50 potency and selectivity of Navi.7 over Navi.5. In particular, a 4-phenyl substitution increases Navi.7 inhibition, particularly, where the 4-phenyl substituent is substantially orthogonal to the isoxazole ring. Further, atropisomerism around the carbon-carbon bond at the 4-position affords chirality that enhances Navi.7 inhibition and selectivity over Navi.5.
[0024] Compound 100, identified through high-throughput screening (HTS), exhibited potency against Navi .7 (570 nM) but poor selectivity over Navi .5 (1.9 uM) in in vitro testing. Nonetheless, compound 100 showed promising compound profiling in ADME assays (solubility, CYP inhibition, in vitro metabolism). When compound 100 was tested in in vivo mouse automated formalin model studies, it caused a 31.4% reduction in late-phase pain-induced reactions albeit at a relatively high dose (135 mg/kg p.o.). Subsequent systematic SAR studies demonstrated that subtype selectivity (Navi.7 vs. Navi.5) could be improved with methylation of the amide nitrogen, as shown in compound 119 or ortho- substitution on the phenyl ring in the 5-position as in compound 130.
[0025] Compound 124 includes a meta-substitution on the phenyl ring in the 5-position that results in reversing subtype selectivity. Compound 124 was found to be very potent againstNavl .5 (36 nM) as well as Navi.7 (120 nM) again, supporting that subtle structural differences have a profound influence on both potency and selectivity. Similarly, a significant increase in Navi.7 potency was realized when a substitution was introduced at the 4-position of the isoxazole ring as exemplified by the 4-phenyl substitution in compound 106. Installation of a 4-phenyl substituent increased Navi.7 inhibition by almost an order of magnitude (60 nM) over compound 100, yet subtype selectivity remained poor (Navi.5 IC50 = 180 nM).
[0026] At this point we also became intrigued with the structure of 106. We hypothesized that phenyl substitution at the 4-position would induce a conformational bias to avoid steric interactions between the adjacent phenyl ring in the 5-position and ester group in the 3-position. Indeed, density function theory (DFT) calculations at the B3LYP/6-31G level of theory identified the lowest energy conformation of a simplified analog of compound 106 that demonstrated a clear preference for the 4-phenyl substituent to be almost completely orthogonal to isoxazole ring, as shown in FIG. 4. The DFT calculations were confirmed by X-ray crystallography of similar tri- substituted isoxazoles where a phenyl ring in the 4-position is orthogonal to the isoxazole ring.
Example 1:
[0027] Compound 100 was administered using the mouse automated formalin model at a dosage of 135 mg/kg p.o. Dosage was selected based upon projection from mouse exposure at 5 mg/kg. Early phase and late phase tonic events were measured with a Tmax equal to 15 minutes and a Tl/2 equal to 1.1 hours. Table 2, below, and FIG. 3 summarize the results of the test demonstrating the Navi.7 inhibitory activity of compound 100 relative to vehicle.
Table 2
[0028] FIG. 5 outlines the synthesis of 3,4,5-trisubstituted isoxazoles in accordance with the present invention. Compound 106 was synthesized by this synthetic route using phenyl boronic acid. Without being bound by theory, atropisomers around the C-C bond at the 4-position are contemplated with either a single ortho- or meta-substituent. Atropisomeric isoxazoles with simple ortho-substitution from aryl boronic acids that includes ortho-fluorine, -chlorine, -bromine, -methyl, and -methoxy may also be made by this synthetic route.
[0029] Chiral separation of tri substituted atropisomeric isoxazoles may be accomplished by interrupting the synthetic route after the Suzuki cross-coupling reaction to yield two complementary methods for separating the atropisomers as shown in FIG. 5. The methyl ester offers an ideal site for chiral semi-preparative HPLC separation. Alternatively, hydrolysis of the methyl ester to the corresponding carboxylic acid as a handle for separation via classical resolution.

Claims

Claims
1. A sodium gated channel Navi.7 inhibitor, comprising the compound having the formula I, a stereoisomer, enantiomer, atropisomer, mixture of enantiomers, mixture of diastereomers, mixture of atropisomers, or isotopic variant thereof; or pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof:
Wherein R1 is selected from the group of methyl, phenyl, chlorophenyl, dichlorophenyl, fluorophenyl, trifluoromethyl, methoxypehnyl, cyanophenyl, pyridine, furan, and thiophene, and combinations thereof;
Wherein R2 is selected from the group of hydrogen, methyl, trifluoromethyl, halogen, alkynyl, phenyl, amide, methylphenyl, and fluoromethylphenyl, and combinations thereof;
Wherein R3 is selected from the group of hydrogen, keto, thioketo, and combinations thereof; Wherein R4 is selected from the group of 1-10 carbon branched or straight chain alkyl, hydroxyalky, cyclic, heterocyclic, sulfide, aldehyde, phenyl, and combinations thereof; and Wherein R5 is selected from the group of hydrogen, methyl, and saturated or unsaturated cycloalkanes having 3-6 carbon atoms.
2. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein the compound further comprises:
3. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein the compound further comprises:
4. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein the compound further comprises
5. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein the compound further comprises:
6. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein the compound exhibits Navi .7 selectivity that is at least two times greater than its selectivity for Navi .5.
7. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein the compound exhibits Navi .7 selectivity that is at least three times greater than its selectivity for Navi .5.
8. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein the R2 substituent group exhibits atropisomerism relative to the the R1 substituent group.
9. The sodium gated channel Navi.7 inhibitor of Claim 1, wherein R1 is a phenyl group.
10. The sodium gated channel Navi.7 inhibitor of Claim 9, wherein the phenyl group further has at least one subsituent group selected from the group of methyl, methoxy, cyano, chloro, dichloro, fluoro and trifluoromethyl.
11. The sodium gated channel Navi.7 inhibitor of Claim 10, wherein the at least one substiuent group is present at one or or both of the ortho or meta positions of the phenyl group.
12. A method of inhibiting Navi .7, comprising the step of administering to a patient in need thereof a pharmaceutically acceptable amount of a compound having the formula I, a stereoisomer, enantiomer, atropisomer, mixture of enantiomers, mixture of diastereomers, mixture of atropisomers, or isotopic variant thereof; or pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof:
Wherein R1 is selected from the group of methyl, phenyl, chlorophenyl, dichlorophenyl, fluorophenyl, trifluoromethyl, methoxypehnyl, cyanophenyl, pyridine, furan, and thiophene, and combinations thereof;
Wherein R2 is selected from the group of hydrogen, methyl, trifluoromethyl, halogen, alkynyl, phenyl, amide, methylphenyl, and fluoromethylphenyl, and combinations thereof;
Wherein R3 is selected from the group of hydrogen, keto, thioketo, and combinations thereof; Wherein R4 is selected from the group of 1-10 carbon branched or straight chain alkyl, hydroxyalky, cyclic, heterocyclic, sulfide, aldehyde, phenyl, and combinations thereof; and Wherein R5 is selected from the group of hydrogen, methyl, and saturated or unsaturated cycloalkanes having 3-6 carbon atoms.
13. The method of Claim 12, wherein the administered compound further comprises:
14. The method of Claim 12, wherein the administered compound further comprises:
15. The method of Claim 12, wherein the administered compound further comprises:
16. The method of Claim 12, wherein the administered compound further comprises:
17. The method of Claim 12, wherein the administered compound exhibits Navi.7 selectivity that is at least two times greater than its selectivity for Navi .5.
18. The method of Claim 12, wherein the administered compound exhibits Navi.7 selectivity that is at least three times greater than its selectivity for Navi .5.
19. The method of Claim 12, wherein the R2 substituent group in the adminstered compound exhibits atropisomerism relative to the the R1 substituent group.
20. The method of Claim 12, wherein R1 is a phenyl group.
21. The method of Claim 20, wherein, wherein the phenyl group further has at least one subsituent group selected from the group of methyl, methoxy, cyano, chloro, dichloro, fluoro and trifluorom ethyl.
22. The method of Claim 21, wherein the at least one substiuent group is present at one or or both of the ortho or meta positions of the phenyl group.
EP21767873.9A 2020-03-13 2021-03-12 Substituted isoxazoles as selective nav1.7 inhibitors for pain treatment and method of pain treatment Pending EP4117658A4 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202062989186P 2020-03-13 2020-03-13
PCT/US2021/022187 WO2021183937A1 (en) 2020-03-13 2021-03-12 Substituted isoxazoles as selective nav1.7 inhibitors for pain treatment and method of pain treatment

Publications (2)

Publication Number Publication Date
EP4117658A1 true EP4117658A1 (en) 2023-01-18
EP4117658A4 EP4117658A4 (en) 2024-05-15

Family

ID=77670919

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21767873.9A Pending EP4117658A4 (en) 2020-03-13 2021-03-12 Substituted isoxazoles as selective nav1.7 inhibitors for pain treatment and method of pain treatment

Country Status (3)

Country Link
EP (1) EP4117658A4 (en)
KR (1) KR20220166810A (en)
WO (1) WO2021183937A1 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0418667B1 (en) * 1989-09-22 1995-08-16 BASF Aktiengesellschaft Carboxylic acid amides
EP2306837B2 (en) * 2008-07-09 2023-10-25 Basf Se Pesticidal active mixtures comprising isoxazoline compounds i
EP3193610A4 (en) * 2014-09-09 2018-04-04 Chromocell Corporation Selective nav1.7 inhibitors for the treatment of diabetes
WO2017133931A1 (en) * 2016-02-04 2017-08-10 Nestec S.A. In vitro production of pancreatic beta cells

Also Published As

Publication number Publication date
EP4117658A4 (en) 2024-05-15
WO2021183937A1 (en) 2021-09-16
KR20220166810A (en) 2022-12-19

Similar Documents

Publication Publication Date Title
EP1908753B1 (en) Novel heterocyclidene acetamide derivative
CN109415336A (en) MDM2 protein degradation agent
KR20140008297A (en) Deuterated analogs of pridopidine useful as dopaminergic stabilizers
Chaudhari et al. Synthesis and pharmacological evaluation of novel N-aryl-3, 4-dihydro-1′ H-spiro [chromene-2, 4′-piperidine]-1′-carboxamides as TRPM8 antagonists
KR20190111080A (en) Bis-heteroaryl Derivatives As Regulators of Protein Aggregation
WO2012116278A1 (en) Cannabinoid receptor modulators
Cevher et al. Discovery of Michael acceptor containing 1, 4-dihydropyridines as first covalent inhibitors of L-/T-type calcium channels
Wichur et al. Development and crystallography-aided SAR studies of multifunctional BuChE inhibitors and 5-HT6R antagonists with β-amyloid anti-aggregation properties
TW201643143A (en) Compound for inhibiting neuronal cell death induced by oxidative stress
US9526721B2 (en) Tetrahydroprotoberbine compounds and uses thereof in the treatment of neurological, psychiatric and neurodegenerative diseases
Xu et al. Discovery of novel brain-penetrant GluN2B NMDAR antagonists via pharmacophore-merging strategy as anti-stroke therapeutic agents
JP2018523644A (en) Selected amides of γ-hydroxybutyric acid and their use in the treatment of alcohol abuse
Gitto et al. Synthesis and Structure–Active Relationship of 1-Aryl-6, 7-dimethoxy-1, 2, 3, 4-tetrahydroisoquinoline Anticonvulsants
EP4117658A1 (en) Substituted isoxazoles as selective nav1.7 inhibitors for pain treatment and method of pain treatment
CA2792918A1 (en) Benzazepine compound
JP6430015B2 (en) A novel chromone oxime derivative and its use as an allosteric modulator of metabotropic glutamate receptors
Malathi et al. Design, synthesis, and pharmacological evaluation of [1, 3] dioxolo-chromeno [2, 3-b] pyridines as anti-seizure agents
CA2888024C (en) Fluorine substituted cyclic amine compounds and preparation methods, pharmaceutical compositions, and uses thereof
Nam et al. Discovery and biological evaluation of tetrahydrothieno [2, 3-c] pyridine derivatives as selective metabotropic glutamate receptor 1 antagonists for the potential treatment of neuropathic pain
CN115515682B (en) Compositions for treating neurodegenerative and mitochondrial diseases and methods of use thereof
Lenkowski et al. Block of human NaV1. 5 sodium channels by novel α-hydroxyphenylamide analogues of phenytoin
Porter et al. An anthrone-based Kv7. 2/7.3 channel blocker with improved properties for the investigation of psychiatric and neurodegenerative disorders
BR112016009918B1 (en) COMPOUND HIGHER INHIBITION OF PROTEIN KINASE G ACTIVITY, ITS USE AND METHOD OF PREPARATION, AND PHARMACEUTICAL COMPOSITION
Zhou et al. Structure-based design of novel G-Protein-Coupled receptor TAAR1 agonists as potential antipsychotic drug candidates
Gao et al. Discovery of a potent, selective, and orally bioavailable histamine H3 receptor antagonist SAR110068 for the treatment of sleep–wake disorders

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20221013

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

RIN1 Information on inventor provided before grant (corrected)

Inventor name: NEFF, ROBYNNE

Inventor name: DIBRELL, SARA

Inventor name: FRANZ, DOUG

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20240415

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 261/06 20060101ALI20240409BHEP

Ipc: A61K 31/42 20060101ALI20240409BHEP

Ipc: A61K 31/4155 20060101ALI20240409BHEP

Ipc: A61K 31/41 20060101AFI20240409BHEP